Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells

<p>Abstract</p> <p>Background</p> <p>Neurotensin has been found to promote colon carcinogenesis in rats and mice, and proliferation of human colon carcinoma cell lines, but the mechanisms involved are not clear. We have examined signalling pathways activated by neuroten...

Full description

Bibliographic Details
Main Authors: Dajani Olav, Dawood Mona, Ødegård John, Aasrum Monica, Tveteraas Ingun H, Müller Kristin M, Christoffersen Thoralf, Sandnes Dagny L
Format: Article
Language:English
Published: BMC 2011-10-01
Series:BMC Cancer
Online Access:http://www.biomedcentral.com/1471-2407/11/421
_version_ 1811279344615555072
author Dajani Olav
Dawood Mona
Ødegård John
Aasrum Monica
Tveteraas Ingun H
Müller Kristin M
Christoffersen Thoralf
Sandnes Dagny L
author_facet Dajani Olav
Dawood Mona
Ødegård John
Aasrum Monica
Tveteraas Ingun H
Müller Kristin M
Christoffersen Thoralf
Sandnes Dagny L
author_sort Dajani Olav
collection DOAJ
description <p>Abstract</p> <p>Background</p> <p>Neurotensin has been found to promote colon carcinogenesis in rats and mice, and proliferation of human colon carcinoma cell lines, but the mechanisms involved are not clear. We have examined signalling pathways activated by neurotensin in colorectal and pancreatic carcinoma cells.</p> <p>Methods</p> <p>Colon carcinoma cell lines HCT116 and HT29 and pancreatic adenocarcinoma cell line Panc-1 were cultured and stimulated with neurotensin or epidermal growth factor (EGF). DNA synthesis was determined by incorporation of radiolabelled thymidine into DNA. Levels and phosphorylation of proteins in signalling pathways were assessed by Western blotting.</p> <p>Results</p> <p>Neurotensin stimulated the phosphorylation of both extracellular signal-regulated kinase (ERK) and Akt in all three cell lines, but apparently did so through different pathways. In Panc-1 cells, neurotensin-induced phosphorylation of ERK, but not Akt, was dependent on protein kinase C (PKC), whereas an inhibitor of the β-isoform of phosphoinositide 3-kinase (PI3K), TGX221, abolished neurotensin-induced Akt phosphorylation in these cells, and there was no evidence of EGF receptor (EGFR) transactivation. In HT29 cells, in contrast, the EGFR tyrosine kinase inhibitor gefitinib blocked neurotensin-stimulated phosphorylation of both ERK and Akt, indicating transactivation of EGFR, independently of PKC. In HCT116 cells, neurotensin induced both a PKC-dependent phosphorylation of ERK and a metalloproteinase-mediated transactivation of EGFR that was associated with a gefitinib-sensitive phosphorylation of the downstream adaptor protein Shc. The activation of Akt was also inhibited by gefitinib, but only partly, suggesting a mechanism in addition to EGFR transactivation. Inhibition of PKC blocked neurotensin-induced DNA synthesis in HCT116 cells.</p> <p>Conclusions</p> <p>While acting predominantly through PKC in Panc-1 cells and via EGFR transactivation in HT29 cells, neurotensin used both these pathways in HCT116 cells. In these cells, neurotensin-induced activation of ERK and stimulation of DNA synthesis was PKC-dependent, whereas activation of the PI3K/Akt pathway was mediated by stimulation of metalloproteinases and subsequent transactivation of the EGFR. Thus, the data show that the signalling mechanisms mediating the effects of neurotensin involve multiple pathways and are cell-dependent.</p>
first_indexed 2024-04-13T00:51:44Z
format Article
id doaj.art-228ebca46041464f81113a6785935e28
institution Directory Open Access Journal
issn 1471-2407
language English
last_indexed 2024-04-13T00:51:44Z
publishDate 2011-10-01
publisher BMC
record_format Article
series BMC Cancer
spelling doaj.art-228ebca46041464f81113a6785935e282022-12-22T03:09:50ZengBMCBMC Cancer1471-24072011-10-0111142110.1186/1471-2407-11-421Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cellsDajani OlavDawood MonaØdegård JohnAasrum MonicaTveteraas Ingun HMüller Kristin MChristoffersen ThoralfSandnes Dagny L<p>Abstract</p> <p>Background</p> <p>Neurotensin has been found to promote colon carcinogenesis in rats and mice, and proliferation of human colon carcinoma cell lines, but the mechanisms involved are not clear. We have examined signalling pathways activated by neurotensin in colorectal and pancreatic carcinoma cells.</p> <p>Methods</p> <p>Colon carcinoma cell lines HCT116 and HT29 and pancreatic adenocarcinoma cell line Panc-1 were cultured and stimulated with neurotensin or epidermal growth factor (EGF). DNA synthesis was determined by incorporation of radiolabelled thymidine into DNA. Levels and phosphorylation of proteins in signalling pathways were assessed by Western blotting.</p> <p>Results</p> <p>Neurotensin stimulated the phosphorylation of both extracellular signal-regulated kinase (ERK) and Akt in all three cell lines, but apparently did so through different pathways. In Panc-1 cells, neurotensin-induced phosphorylation of ERK, but not Akt, was dependent on protein kinase C (PKC), whereas an inhibitor of the β-isoform of phosphoinositide 3-kinase (PI3K), TGX221, abolished neurotensin-induced Akt phosphorylation in these cells, and there was no evidence of EGF receptor (EGFR) transactivation. In HT29 cells, in contrast, the EGFR tyrosine kinase inhibitor gefitinib blocked neurotensin-stimulated phosphorylation of both ERK and Akt, indicating transactivation of EGFR, independently of PKC. In HCT116 cells, neurotensin induced both a PKC-dependent phosphorylation of ERK and a metalloproteinase-mediated transactivation of EGFR that was associated with a gefitinib-sensitive phosphorylation of the downstream adaptor protein Shc. The activation of Akt was also inhibited by gefitinib, but only partly, suggesting a mechanism in addition to EGFR transactivation. Inhibition of PKC blocked neurotensin-induced DNA synthesis in HCT116 cells.</p> <p>Conclusions</p> <p>While acting predominantly through PKC in Panc-1 cells and via EGFR transactivation in HT29 cells, neurotensin used both these pathways in HCT116 cells. In these cells, neurotensin-induced activation of ERK and stimulation of DNA synthesis was PKC-dependent, whereas activation of the PI3K/Akt pathway was mediated by stimulation of metalloproteinases and subsequent transactivation of the EGFR. Thus, the data show that the signalling mechanisms mediating the effects of neurotensin involve multiple pathways and are cell-dependent.</p>http://www.biomedcentral.com/1471-2407/11/421
spellingShingle Dajani Olav
Dawood Mona
Ødegård John
Aasrum Monica
Tveteraas Ingun H
Müller Kristin M
Christoffersen Thoralf
Sandnes Dagny L
Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
BMC Cancer
title Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
title_full Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
title_fullStr Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
title_full_unstemmed Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
title_short Role of protein kinase C and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
title_sort role of protein kinase c and epidermal growth factor receptor signalling in growth stimulation by neurotensin in colon carcinoma cells
url http://www.biomedcentral.com/1471-2407/11/421
work_keys_str_mv AT dajaniolav roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT dawoodmona roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT ødegardjohn roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT aasrummonica roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT tveteraasingunh roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT mullerkristinm roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT christoffersenthoralf roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells
AT sandnesdagnyl roleofproteinkinasecandepidermalgrowthfactorreceptorsignallingingrowthstimulationbyneurotensinincoloncarcinomacells